Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David Briere is active.

Publication


Featured researches published by David Briere.


Molecular Cancer Therapeutics | 2010

PF-03814735, an Orally Bioavailable Small Molecule Aurora Kinase Inhibitor for Cancer Therapy

Jitesh P. Jani; Vincent Bernardo; Samit Kumar Bhattacharya; David Briere; Bruce D. Cohen; Kevin Coleman; James G. Christensen; Erling O. Emerson; Amy B. Jakowski; Kenneth E. Hook; Gerrit Los; James D. Moyer; Ingrid Pruimboom-Brees; Leslie R. Pustilnik; Ann Marie Rossi; Stefan J. Steyn; Chunyan Su; Konstantinos Tsaparikos; Donn G. Wishka; Kwansik Yoon; John Jakubczak

The Aurora family of highly related serine/threonine kinases plays a key role in the regulation of mitosis. Aurora1 and Aurora2 play important but distinct roles in the G2 and M phases of the cell cycle and are essential for proper chromosome segregation and cell division. Overexpression and amplification of Aurora2 have been reported in different tumor types, including breast, colon, pancreatic, ovarian, and gastric cancer. PF-03814735 is a novel, potent, orally bioavailable, reversible inhibitor of both Aurora1 and Aurora2 kinases that is currently in phase I clinical trials for the treatment of advanced solid tumors. In intact cells, the inhibitory activity of PF-03814735 on the Aurora1 and Aurora2 kinases reduces levels of phospho-Aurora1, phosphohistone H3, and phospho-Aurora2. PF-03814735 produces a block in cytokinesis, resulting in inhibition of cell proliferation and the formation of polyploid multinucleated cells. Although PF-03814735 produces significant inhibition of several other protein kinases, the predominant biochemical effects in cellular assays are consistent with inhibition of Aurora kinases. Once-daily oral administration of PF-03814735 to mice bearing human xenograft tumors produces a reduction in phosphohistone H3 in tumors at doses that are tolerable and that result in significant inhibition of tumor growth. The combination of PF-03814735 and docetaxel in xenograft mouse tumor models shows additive tumor growth inhibition. These results support the clinical evaluation of PF-03814735 in cancer patients. Mol Cancer Ther; 9(4); 883–94. ©2010 AACR.


Molecular Cancer Therapeutics | 2013

A Novel Antiandrogen, Compound 30, Suppresses Castration-Resistant and MDV3100-Resistant Prostate Cancer Growth In Vitro and In Vivo

Hidetoshi Kuruma; Hiroaki Matsumoto; Masaki Shiota; Jennifer L. Bishop; Francois Lamoureux; Christian Thomas; David Briere; Gerrit Los; Martin E. Gleave; Andrea Fanjul; Amina Zoubeidi

Resistance to antiandrogen drugs, like MDV3100, occurs in patients with castration-resistant prostate cancer (CRPC). Thus, preventing or treating antiandrogen resistance is a major clinical challenge. We identified a novel antiandrogen, Compound 30, and compared its efficacy with MDV3100. We found that Compound 30 inhibits androgen receptor (AR) activity in LNCaP cells, C4-2 cells, as well as MDV3100-resistant cell lines. Compared with MDV3100, Compound 30 treatment induces greater reduction in AR, prostate-specific antigen (PSA), and AR transcriptional activity, and prevents AR nuclear translocation in AR-sensitive LNCaP cells. Compound 30 has antiproliferative effects in LNCaP cells, in castrate-resistant C4-2 cells, and those resistant to MDV3100. Compound 30 was equally as effective as MDV3100 in reducing tumor volume and PSA in vivo. More importantly, Compound 30 is effective at inhibiting AR activity in MDV3100-resistant cell lines and significantly prevented tumor growth and PSA increases in mice bearing MDV3100-resistant xenografts. Together, our data show that Compound 30 strongly inhibited AR activity and suppressed castration-resistant LNCaP growth as well as MDV3100-resistant cell growth in vitro and in vivo. These data provide a preclinical proof-of-principle that Compound 30 could be a promising next generation anti-AR agent, especially in the context of antiandrogen-resistant tumors. Mol Cancer Ther; 12(5); 567–76. ©2013 AACR.


Journal of Medicinal Chemistry | 2010

Design of selective, ATP-competitive inhibitors of Akt.

Kevin Daniel Freeman-Cook; Christopher Autry; Gary Borzillo; Deborah Gordon; Elsa G. Barbacci-Tobin; Vincent Bernardo; David Briere; Tracey Clark; Matthew Corbett; John Jakubczak; Shefali Kakar; Elizabeth Knauth; Blaise Lippa; Michael Joseph Luzzio; Mahmoud N. Mansour; Gary J. Martinelli; Matthew A. Marx; Kendra Louise Nelson; Jayvardhan Pandit; Francis Rajamohan; Shaughnessy Robinson; Chakrapani Subramanyam; Liuqing Wei; Martin James Wythes; Joel Morris

This paper describes the design and synthesis of novel, ATP-competitive Akt inhibitors from an elaborated 3-aminopyrrolidine scaffold. Key findings include the discovery of an initial lead that was modestly selective and medicinal chemistry optimization of that lead to provide more selective analogues. Analysis of the data suggested that highly lipophilic analogues would likely suffer from poor overall properties. Central to the discussion is the concept of optimization of lipophilic efficiency and the ability to balance overall druglike propeties with the careful control of lipophilicity in the lead series. Discovery of the nonracemic amide series and subsequent modification produced an advanced analogue that performed well in advanced preclinical assays, including xenograft tumor growth inhibition studies, and this analogue was nominated for clinical development.


BMC Genomics | 2012

Dose-dependent effects of small-molecule antagonists on the genomic landscape of androgen receptor binding

Zhou Zhu; Manli Shi; Wenyue Hu; Heather Estrella; Jon Engebretsen; Tim Nichols; David Briere; Natilie Hosea; Gerrit Los; Paul A. Rejto; Andrea Fanjul

BackgroundThe androgen receptor plays a critical role throughout the progression of prostate cancer and is an important drug target for this disease. While chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-Seq) is becoming an essential tool for studying transcription and chromatin modification factors, it has rarely been employed in the context of drug discovery.ResultsHere we report changes in the genome-wide AR binding landscape due to dose-dependent inhibition by drug-like small molecules using ChIP-Seq. Integration of sequence analysis, transcriptome profiling, cell viability assays and xenograft tumor growth inhibition studies enabled us to establish a direct cistrome-activity relationship for two novel potent AR antagonists. By selectively occupying the strongest binding sites, AR signaling remains active even when androgen levels are low, as is characteristic of first-line androgen ablation therapy. Coupled cistrome and transcriptome profiling upon small molecule antagonism led to the identification of a core set of AR direct effector genes that are most likely to mediate the activities of targeted agents: unbiased pathway mapping revealed that AR is a key modulator of steroid metabolism by forming a tightly controlled feedback loop with other nuclear receptor family members and this oncogenic effect can be relieved by antagonist treatment. Furthermore, we found that AR also has an extensive role in negative gene regulation, with estrogen (related) receptor likely mediating its function as a transcriptional repressor.ConclusionsOur study provides a global and dynamic view of AR’s regulatory program upon antagonism, which may serve as a molecular basis for deciphering and developing AR therapeutics.


Cancer Research | 2016

Abstract 2642: Evaluation of the mechanism of MET-dependent cellular transformation and potent cytoreductive activity of MGCD265 in novel MET exon 14 mutation positive cancer models

Lars D. Engstrom; Ruth W. Tang; David Briere; Harrah Chiang; Peter Olson; James G. Christensen

MET splice site mutations that result in the deletion of exon 14 (METex14del) are implicated as oncogenic drivers in a subset of non-small cell lung cancer (NSCLC). MET exon 14 contains the Y1003 CBL ubiquitin ligase regulatory binding site that normally mediates CBL-dependent MET degradation and signal attenuation. Deletion of this exon results in sustained activation of MET and its downstream signaling pathways. The diverse splice site mutations leading to exon 14 skipping comprise a unique and unprecedented class of RTK activating mutations and the molecular mechanism by which these genetic alterations transform cancer cells is not fully understood. One major challenge in understanding the utility of MET inhibition of the METex14del class has been the lack of available pre-clinical models. In the present study, we generated and characterized multiple METex14del-driven cancer models to study the mechanism of MET-dependent cellular transformation as well as the response to MGCD265, a small molecule inhibitor of MET and AXL. METex14del models were identified via mining patient-derived xenograft (PDX) databases or were engineered using genome editing techniques to generate isogenic pairs of METex14del and WT cell lines. The METex14del cell lines formed increased size and number of colonies in anchorage independent growth assays compared to their WT counterparts. The transformation of METex14del cells was associated with an increase in durable HGF-dependent activation of MET and downstream signaling pathways potentially due to dysregulated MET processing and signaling attenuation. MGCD265 was shown to effectively inhibit this growth and MET-dependent signal transduction in a concentration-dependent manner. When evaluated in the amplified METex14del-driven gastric xenograft model Hs746T, significant tumor regression was observed following MGCD265 treatment. In addition, MGCD265 demonstrated substantial regression of large established tumors, in two novel NSCLC METex14del-positive PDX models. Together, these data confirm METex14del mutations are bona fide oncogenic drivers and sensitive to targeted therapeutics. Moreover, the models described in this study represent a relevant pre-clinical platform to further study receptor hyper-activation and drug action that is clinically actionable. Identification, development, and understanding of METex14del models will likely help further guide precision medicine strategies to treat NSCLC patients harboring these mutations. Citation Format: Lars D. Engstrom, Ruth W. Tang, David M. Briere, Harrah Chiang, Peter Olson, James G. Christensen. Evaluation of the mechanism of MET-dependent cellular transformation and potent cytoreductive activity of MGCD265 in novel MET exon 14 mutation positive cancer models. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2642.


Cancer Research | 2016

Abstract 4021: The class I HDAC inhibitor, mocetinostat, induces expression of PD-L1 and tumor antigen presentation machinery and modifies tumor immune cellular subsets providing a rationale for immune checkpoint inhibitor combinations

David Briere; Niranjan Sudhakar; Lars D. Engstrom; Jill Hallin; Ruth W. Tang; Harrah Chiang; Maryland Rosenfeld-Franklin; Peter Olson; James G. Christensen

Immunotherapy has led to major treatment breakthroughs for a number of cancers including non-small cell lung cancer (NSCLC). Although initial responses to immune checkpoint inhibitors are promising, a significant percentage of patients do not respond or rapidly acquire resistance. Although the mechanisms underlying intrinsic and acquired resistance remain largely unexplained; the expression of programmed cell death-ligand 1 (PD-L1), lack of tumor cell capacity to effectively present neoantigens, and presence of immunosuppressive cellular subsets have been implicated as potential mechanisms. Histone deacetylase (HDAC) inhibitors have emerged as a class of agents that may combat checkpoint inhibitor resistance by reversing immune evasion and eliciting an anti-tumor activity through a multi-faceted immuno-stimulatory mechanism of action. Mocetinostat is a spectrum-selective Class I/IV HDAC inhibitor specifically targeting HDAC-1, -2, -3 and -11. The present studies were designed to explore mocetinostat9s effect as an immune-enhancer and ultimately, to evaluate its potential to be used in combination with immune checkpoint inhibitors (e.g., PD-1/PD-L1 antagonists). Specifically, we assessed mocetinostat9s effect on the expression of various immunomodulatory factors by tumor cells as well as its effect on immune cell sub-populations in the tumor microenvironment in vivo. Mocetinostat elicited a concentration-dependent increase in PD-L1 mRNA expression which translated into increased PD-L1 surface protein expression in a panel of NSCLC cell lines. In addition, mocetinostat elicited a concentration-dependent increase in expression of MHC-class I related polypeptide-related sequence A (MIC-A) and MIC-B, and cluster of differentiation 86 (CD86). Furthermore, mocetinostat induced expression of several human leukocyte antigen (HLA) gene complex family members including HLA-A, -B, -DRA, and -DPA among others. To determine the effect of mocetinostat on systemic and tumor immune cell subpopulations we treated CT26 tumor-bearing mice. Mocetinostat increased splenic CD4-positive T effector cells and tumor mature cytolytic CD8-postive T cells and at the same time decreased tumor FoxP3-positive T regulatory cells and CD11b/Gr1-positive myeloid-derived suppressor cells (MDSC). These data provide evidence that mocetinostat modulates key immune regulators both in tumor cells as well as in relevant immune cell types in the tumor microenvironment and provides strong rationale for combination with immune checkpoint inhibitors. Citation Format: David Briere, Niranjan Sudhakar, Lars Engstrom, Jill Hallin, Ruth Tang, Harrah Chiang, Maryland Rosenfeld-Franklin, Peter Olson, James Christensen. The class I HDAC inhibitor, mocetinostat, induces expression of PD-L1 and tumor antigen presentation machinery and modifies tumor immune cellular subsets providing a rationale for immune checkpoint inhibitor combinations. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4021.


Molecular Cancer Therapeutics | 2009

Abstract B258: Investigating PERK biological pathway using protein/peptide microarrays and SAR with small molecule inhibitors

Hubert Otlik; Sergei Timofeevski; Terri Quenzer; David Briere; Andrea Fanjul; Gordon Alton; Jacques Ermolieff

Accumulation of unfolded proteins in the endoplasmic occurs when the cell is subject to stress caused by various pathological conditions such as hypoxia, viral infection, and glucose depravation. Under such stress the cell will initiate an unfolded protein response (UPR), a protective mechanism that is specifically designed to re‐establish homeostasis and normal endoplasmic reticulum (ER) function. This adaptive mechanism inhibits overall protein translation, but enhances the translation of a small number of key stress response proteins that will clear the ER of unfolded proteins and send them to the cytoplasm for degradation. The UPR is initiated by several proteins such as IRE1a, ATF6a and PERK, the later being a key Ser/Thr protein kinase in UPR signaling. In response to inducers of ER stress, BiP (GRP78) dissociates from the luminal ER domain of PERK, resulting in the oligomerization, autophosphorylation, and activation of PERK which in turn phosphorylates eIF‐2a on Ser51 and Nrf2 (unknown site of phosphorylation). In order to further identify potential substrates for PERK, this enzyme was tested against large protein and peptide microarrays. A significant number of proteins and peptides in the microarrays were found to be phosphorylated by PERK. After additional selection, several of these substrates were further characterized using a microfluidic mobility‐shift assay and submitted to LC/MS analysis to identify the site of phosphorylation by PERK. These newly identified peptide substrates were used to develop a robust biochemical assay for the testing of small molecule inhibitors of PERK activity. A limited SAR was established for a set of compounds against PERK and two other related protein kinases, GCN2 and PKR. One of the most potent and selective PERK inhibitors was found to modulate PERK cellular signaling as evidenced by blocking the phosphorylation of (Ser51)eIF‐2a as judged by Elisa and Western blot analysis. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B258.


Journal of Medicinal Chemistry | 2011

Discovery of aryloxy tetramethylcyclobutanes as novel androgen receptor antagonists.

Chuangxing Guo; Angelica Linton; Susan Kephart; Martha Ornelas; Mason Alan Pairish; Javier Gonzalez; Samantha Greasley; Asako Nagata; Benjamin J. Burke; Martin Paul Edwards; Natilie Hosea; Ping Kang; Wenyue Hu; Jon Engebretsen; David Briere; Manli Shi; Hovik Gukasyan; Paul G. Richardson; Kevin Neil Dack; Toby J. Underwood; Patrick Stephen Johnson; Andrew Morell; Robert Felstead; Hidetoshi Kuruma; Hiroaki Matsimoto; Amina Zoubeidi; Martin Gleave; Gerrit Los; Andrea Fanjul


Cancer Research | 2008

SAR and chemistry of Aurora kinase inhibitors: Discovery of PF-3814735, an oral clinical candidate

Samit Kumar Bhattacharya; Donn G. Wishka; Michael Joseph Luzzio; Vincent Bernardo; David Briere; Tracy Lee Boyden; Mary Campbell; Bruce M. Cohen; Kevin Coleman; Erling Emerson; John Jakubczak; Jitesh P. Jani; Chao Li; Matthew A. Marx; James D. Moyer; Kendra Louise Nelson; Gonghua Pan; Leslie R. Pustilnik; Kristina Rafidi; Ann Marie Rossi; Rick Shepard; Stefanus J. Steyn; Chunyan Su; Shusen Sun; Kostantinos Tsaparikos; Courtney Williams; Jun Xiao; Kwansik Yoon; Xumiao Zhao


Cancer Immunology, Immunotherapy | 2018

The class I/IV HDAC inhibitor mocetinostat increases tumor antigen presentation, decreases immune suppressive cell types and augments checkpoint inhibitor therapy

David Briere; Niranjan Sudhakar; David M. Woods; Jill Hallin; Lars D. Engstrom; Ruth Aranda; Harrah Chiang; Andressa L. Sodre; Peter Olson; Jeffrey S. Weber; James G. Christensen

Collaboration


Dive into the David Briere's collaboration.

Researchain Logo
Decentralizing Knowledge